Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Appl Phys B ; 125(211)2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31920221

ABSTRACT

We present the design of a portable version of our miniaturized laser heterodyne radiometer (mini-LHR) that simultaneously measures methane (CH4) and carbon dioxide (CO2) in the atmospheric column. The mini-LHR fits on a backpack frame, operates autonomously, and requires no infrastructure because it is powered by batteries charged by a folding 30 W solar panel. Similar to our earlier instruments, the mini-LHR is a passive laser heterodyne radiometer that operates by collecting sunlight that has undergone absorption by CH4 and CO2. Within the mini-LHR, sunlight is mixed with light from a distributive feedback (DFB) laser centered at approximately 1.64 µm where both gases have absorption features. The laser scans across these absorption features roughly every minute and the resulting beat signal is collected in the radio frequency (RF). Scans are averaged into half hour and hour data products and analyzed using the Planetary Spectrum Generator (PSG) retrieval to extract column mole fractions. Instrument performance is demonstrated through two deployments at significantly different sites in interior Alaska and Hawaii. The resolving power (λ/∆λ) is greater than 500,000 at 1.64 µm with precisions of better than 20 ppb and 1 ppm for CH4 and CO2, respectively. Because mini-LHR instruments are portable and can be co-located, they can be used to characterize bias between larger, stationary, column observing instruments. In addition, mini-LHRs can be deployed quickly to respond to transient events such as methane leaks or can be used for field studies targeting geographical regions.

2.
Appl Phys B ; 120(4): 609-615, 2015.
Article in English | MEDLINE | ID: mdl-26321796

ABSTRACT

We present column CO2 measurements taken by the passive miniaturized laser heterodyne radiometer (Mini-LHR) at 1611.51 nm at the Mauna Loa Observatory in Hawaii. The Mini-LHR was operated autonomously, during the month of May 2013 at this site, working in tandem with an AERONET sun photometer that measures aerosol optical depth at 15-min intervals during daylight hours. Laser heterodyne radiometry has been used since the 1970s to measure atmospheric gases such as ozone, water vapor, methane, ammonia, chlorine monoxide, and nitrous oxide. This iteration of the technology utilizes distributed feedback lasers to produce a low-cost, small, portable sensor that has potential for global deployment. Applications of this instrument include supplementation of existing monitoring networks to provide denser global coverage, providing validation for larger satellite missions, and targeting regions of carbon flux uncertainty. Also presented here are preliminary retrieval analysis and the performance analysis that demonstrate that the Mini-LHR responds extremely well to changes in the atmospheric absorption.

3.
J Cell Physiol ; 185(3): 416-24, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11056012

ABSTRACT

A prostatic smooth muscle cell line (PSMC1) was established from the dorsolateral prostate of p53 null mice. The cell line is nontumorigenic when inoculated subcutaneously, under the renal capsule or intraprostatically in syngeneic mice. These cells express alpha-smooth muscle actin (alpha-SMA), indicating their smooth muscle origin, and TGF-beta significantly enhances expression of alpha-SMA. The cells express both androgen receptor (AR) mRNA and protein, and respond mitogenically to physiological concentrations of androgens. PSMC1 cells produce significant amounts of TGF-beta, which stimulates growth by an autocrine mechanism. Dihydrotestosterone (DHT) increases proliferation of PSMC1 cells by promoting TGF-beta secretion. Considering the significant inhibitory effect of TGF-beta on prostatic epithelial cells and its stimulatory effect on the PSMC1 cells, we postulate that TGF-beta produced by prostatic smooth muscle cells may have a paracrine effect on the prostatic epithelium. We also postulate that TGF-beta may be involved in the etiology of benign prostatic hyperplasia (BPH) by stimulating excessive stromal proliferation. Line PSMC1 is the first reported androgen-responsive murine smooth muscle cell line. It will be useful for in vivo and in vitro experiments to study the mechanisms of androgen action on prostatic stroma and for delineating the interactions that occur between prostatic smooth muscle and epithelium that may lead to prostatic diseases such as BPH.


Subject(s)
Androgens/physiology , Autocrine Communication/physiology , Muscle, Smooth/cytology , Muscle, Smooth/physiology , Prostate/cytology , Prostate/physiology , Transforming Growth Factor beta/physiology , Androgens/pharmacology , Animals , Cell Line , Male , Mice , Receptors, Androgen/physiology
4.
J Cell Physiol ; 184(1): 70-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10825235

ABSTRACT

Two prostatic epithelial lines, one of basal origin and one of luminal origin, were established from the dorsolateral prostates of p53 null mice. The cell lines are nontumorigenic when inoculated subcutaneously under the renal capsule or intraprostatically in syngeneic mice. The luminal cell line (PE-L-1) expresses cytokeratins 8 and 18 and the basal cell line (PE-B-1) expresses cytokeratins 5 and 14. The basal cells require serum for growth, whereas the luminal cells grow only in serum-free medium. Both cell lines require the presence of growth factors for optimal growth in culture, with EGF and FGF-2 having the greatest effect on the growth rate. Both lines express androgen receptor (AR) mRNA and protein. Androgen stimulates growth of the basal cell line, indicating that the ARs are functional, whereas growth of the luminal cells is unaffected by androgens. The luminal line is significantly inhibited by exogenous TGF-beta and produces low levels of endogenous TGF-beta. In contrast, the basal cell line produces significant amounts of TGF-beta and its growth is not influenced by this cytokine. Coculture of luminal cells with prostatic smooth muscle cells results in the generation of increased levels of biologically active TGF-beta, indicating a paracrine mechanism of TGF-beta activation that may be involved in the maintenance of normal prostatic function. To our knowledge this is the first report describing both basal and luminal prostatic cell lines from a single inbred animal species and the first indication that prostatic epithelial and stromal cells interact to generate the biologically active form of TGF-beta. These lines will provide an important model for determining basal/luminal interactions in both in vitro and in vivo assays.


Subject(s)
Epithelial Cells/physiology , Growth Substances/pharmacology , Prostate/physiology , Transforming Growth Factor beta/biosynthesis , Animals , Biological Assay , Cell Division/drug effects , Cell Line , Cholera Toxin/pharmacology , Coculture Techniques , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Genes, p53 , Hydrocortisone/pharmacology , Insulin/pharmacology , Keratins/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Prostate/cytology , Prostate/drug effects , Receptors, Androgen/analysis , Receptors, Androgen/genetics , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology
5.
J Cell Physiol ; 180(2): 225-35, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10395292

ABSTRACT

The glycosylphosphatidylinositol (GPI)-anchored, multifunctional receptor for the serine proteinase, urokinase plasminogen activator (uPAR, CD87), regulates plasminogen activation and cell migration, adhesion, and proliferation. uPAR occurs in functionally distinct, membrane-anchored and soluble isoforms (s-uPAR) in vitro and in vivo. Recent evidence indicates that s-uPAR present in the circulation of cancer patients correlates with tumor malignancy and represents a valuable prognostic marker in certain types of cancer. We have therefore analyzed the mechanism of uPAR shedding in vitro. We present evidence that uPAR is actively released from ovarian cancer cells since the rate of receptor shedding did not correlate with uPAR expression. While s-uPAR was derived from the cell surface, it lacked the hydrophobic portion of the GPI moiety indicating anchor cleavage. We show that uPAR release is catalyzed by cellular GPI-specific phospholipase D (GPI-PLD), an enzyme cleaving the GPI anchor of the receptor. Thus, recombinant GPI-PLD expression increased receptor release up to fourfold. Conversely, a 40% reduction in GPI-PLD activity by GPI-PLD antisense mRNA expression inhibited uPAR release by more than 60%. We found that GPI-PLD also regulated uPAR expression, possibly by releasing a GPI-anchored growth factor. Our data suggest that cellular GPI-PLD might be involved in the generation of circulating prognostic markers in cancer and possibly regulate the function of GPI-anchored proteins by generating functionally distinct, soluble counterparts.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Phospholipase D/metabolism , Plasminogen Activators/metabolism , Receptors, Cell Surface/metabolism , Antisense Elements (Genetics) , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycosylphosphatidylinositols/genetics , Humans , Hydrolysis , Membrane Proteins/metabolism , Ovarian Neoplasms , Phospholipase D/genetics , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/enzymology
6.
Am J Pathol ; 150(1): 359-69, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9006351

ABSTRACT

Urokinase-type plasminogen activator (uPA) is thought to be an important mediator in the proteolytic degradation of extracellular matrix components observed in a wide variety of normal physiological and pathological conditions. However, the phenotype of a recently developed strain of urokinase-deficient (uPA-/-) mice appears to be normal when maintained under ideal nonstressful conditions. We report an outbreak of botryomycosis, an unusual staphylococcal infection, in a colony of uPA-deficient mice. A detailed histological examination of these uPA-deficient animals also revealed a variety of previously unreported phenotypic abnormalities such as pleuritis and the effacement of lymphoid follicles in the regional lymph nodes and spleen. Additional phenotypic abnormalities such as dystrophic calcifications and rectal prolapse were also observed in the uPA-deficient population. These abnormalities were also noted in ostensibly healthy uPA-deficient animals. Botryomycosis did not affect a colony of wild-type (uPA+/+) animals maintained concurrently under identical conditions in the same room. The peculiar predisposition of the uPA-deficient animals to this rare bacterial infection and the development of phenotypic abnormalities associated with the targeted disruption the uPA gene suggests that uPA contributes significantly to the cutaneous microenvironment and is additional evidence of the extensive involvement of the plasminogen activators in mammalian physiology.


Subject(s)
Lymphoid Tissue/pathology , Pleurisy/genetics , Staphylococcal Skin Infections/genetics , Urokinase-Type Plasminogen Activator/deficiency , Urokinase-Type Plasminogen Activator/genetics , Abscess/pathology , Animals , Cell Movement , Disease Susceptibility , Lung/pathology , Lymph Nodes/pathology , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Pleurisy/enzymology , Pleurisy/pathology , Rectal Prolapse/pathology , Spleen/pathology , Staphylococcal Skin Infections/enzymology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/isolation & purification
7.
Anticancer Res ; 16(5A): 2513-6, 1996.
Article in English | MEDLINE | ID: mdl-8917344

ABSTRACT

A number of proteins are found attached to the plasma membrane of mammalian cells by a glycosylphosphatidylinositol (GPI) anchor that can be cleaved by GPI specific phospholipase D (GPI-PLD). There are no known specific inhibitors of GPI-PLD. We examined some inhibitors of phosphatidylinositol specific phospholipase C (PI-PLC) for their ability to inhibit human serum and human bone marrow cell GPI-PLD. Azo analogues of suramin were found to be potent inhibitors of GPI-PLD. One compound had an IC50 of 3.7 microM that was 10-fold lower than the IC50 required to inhibit PI-PLC. The azo suramin analogues inhibited cancer cell growth at concentrations similar to those required to inhibit GPI-PLD, and below concentrations required to inhibit growth factor binding. It is possible that inhibition of cell growth might be related to the ability of the compounds to inhibit GPI-PLD.


Subject(s)
Phospholipase D/antagonists & inhibitors , Suramin/pharmacology , Cell Division/drug effects , HT29 Cells/drug effects , Humans , Suramin/analogs & derivatives , Tumor Cells, Cultured/drug effects
8.
Cancer Res ; 56(15): 3597-604, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8758932

ABSTRACT

Evidence suggests that the plasminogen activators (PAs), in particular urokinase-type PA (uPA), play a pivotal role in tumor invasion and metastasis. We studied the contribution of the PAs to the malignant phenotype through the chemical induction of melanocytic neoplasms in uPA-deficient mice. Primary tumors were induced and promoted concurrently in 35 uPA-/- deficient and 35 uPA+/+ wild-type mice using a single application of 7,12-dimethylbenz(a)anthracene followed by repetitive applications of croton oil. Animals were sacrificed at 60-day intervals for 1 year. At necropsy, the four largest pigmented lesions in each animal were excised, characterized histologically, and evaluated microscopically for evidence of invasion. The regional lymph nodes, lungs, and solid abdominal visceral organs were sectioned and examined microscopically for evidence of metastatic disease. Cellular blue nevi were induced in 100% of uPA-/- and uPA+/+ promoted animals. Although a reduction in the radial and vertical progression of these lesions was noted in the uPA-deficient mice compared with the wild-type group, more than 95% of cellular blue nevi induced in both groups of animals invaded the underlying tissues. These lesions did not metastasize to the regional lymph nodes. Malignant melanoma arose in 5 of 35 (14.3%) of promoted wild-type mice. These tumors were locally aggressive, produced tissue-type PA, but were not metastatic to the regional nodes, lungs, or abdominal viscera. These results indicate that the invasive capability of melanocytic lesions may depend more on tissue-type PA than uPA activity. No melanomas were induced in the uPA-/- mice. The resistance of the uPA -/- strain to melanoma induction suggests that uPA contributes to malignant progression. We propose that the absence of uPA negatively affects tumorigenesis by decreasing the liberation and availability of growth factors such as basic fibroblast growth factor.


Subject(s)
Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Nevus, Blue/enzymology , Nevus, Blue/pathology , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Urokinase-Type Plasminogen Activator/deficiency , Animals , Cell Division/physiology , Cell Transformation, Neoplastic , Disease Progression , Lymphatic Metastasis , Melanocytes/enzymology , Melanocytes/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism
9.
Blood ; 87(3): 1056-63, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8562930

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by delayed senescence and slow accumulation of monoclonal, small lymphocytes. Basic fibroblast growth factor (bFGF) is a pleiotropic cytokine that plays a role in hematopoiesis and apoptosis. Elevated bFGF levels have been detected in urine from patients with a variety of neoplastic diseases including various leukemias; however, the cellular source of the bFGF has not been determined. In this study, the intracellular bFGF level in lymphocytes of 36 patients with B-CLL and 15 normal donors was determined using an enzyme-linked immunoassay. In cells derived from patients with high-risk disease, the median level of intracellular bFGF was 381.5 pg/2 x 10(5) cells, compared with a median of 90.5 pg/2 x 10(5) cells in patients with intermediate disease. In patients with low-risk disease, the median bFGF level was 4.9 pg/2 x 10(5) cells, and in normal controls, it was 6.0 pg/2 x 10(5) cells. The difference in the bFGF levels was significant for the comparison between low- and intermediate-risk (P = .00119), low- and high-risk (P < .0001), and intermediate- and high-risk disease (P = .0001). Immunofluorescent stains of peripheral blood mononuclear cells confirmed CLL lymphocytes as a cellular source of bFGF. To evaluate the potential contribution of elevated intracellular bFGF levels to the phenotype of CLL cells, leukemic cells were cultured in vitro with an apoptotic stimulus (fludarabine). CLL cells with high intracellular levels of bFGF appeared to be more resistant to fludarabine treatment. The addition of bFGF to fludarabine-treated CLL cells resulted in a delay of apoptosis and prolonged survival. These data suggest that bFGF may contribute to the resistance of CLL cells to an apoptotic stimulus.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 2/analysis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/analysis , Vidarabine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Drug Resistance, Neoplasm , Female , Fibroblast Growth Factor 2/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Staging , Tumor Cells, Cultured/drug effects , Vidarabine/pharmacology
10.
Blood ; 83(7): 1808-12, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8142649

ABSTRACT

Basic fibroblast growth factor (bFGF) and transforming growth factor-beta 1 (TGF-beta) have both been shown to act on hematopoietic progenitor cells. bFGF is a hematopoietic cytokine that acts on progenitor cells in concert with other cytokines to promote their proliferation. TGF-beta induces erythroid differentiation in K562 cells. To determine whether bFGF might act on progenitor cells by antagonizing the effects of cytokines that induce differentiation, we determined the effects of bFGF on the TGF-beta-mediated induction of hemoglobin synthesis in K562 cells. bFGF antagonized the TGF-beta-mediated induction of hemoglobin in a dose-dependent manner, with 0.1 ng/mL bFGF inhibiting hemoglobin induction by 40% and 10 ng/mL bFGF completely abrogating hemoglobin production. bFGF was most effective at antagonizing the TGF-beta-mediated induction of hemoglobin if it and TGF-beta were added simultaneously to K562 cells, but delayed addition of bFGF to TGF-beta-treated cultures still resulted in significant inhibition of hemoglobin synthesis. The inhibitory effects of bFGF on hemoglobin production were fully reversible, showing that bFGF did not permanently alter the phenotype of K562 cells. The hemin-mediated induction of hemoglobin synthesis in K562 cells was only partially negated by bFGF. bFGF also diminished the expression of glycophorin A on the surface of K562 cells. These results indicate that bFGF might increase progenitor/stem cell numbers by antagonizing the effects of cytokines that induce differentiation, thereby increasing the pool of proliferating progenitor/stem cells.


Subject(s)
Erythroid Precursor Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Leukemia, Erythroblastic, Acute/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Cell Differentiation/drug effects , Erythroid Precursor Cells/physiology , Hemoglobins/biosynthesis , Humans , Tumor Cells, Cultured
11.
Blood ; 83(8): 2115-25, 1994 Apr 15.
Article in English | MEDLINE | ID: mdl-8161780

ABSTRACT

Basic fibroblast growth factor (bFGF) is a hematopoietic cytokine that stimulates stromal and stem cell growth. It binds to a glycosylphosphatidylinositol (GPI)-anchored heparan sulfate proteoglycan on human bone marrow (BM) stromal cells. The bFGF-proteoglycan complex is biologically active and is released by addition of exogenous phosphatidylinositol-specific phospholipase C. In this study, we show the presence of an endogenous GPI-specific phospholipase D (GPI-PLD) that releases the bFGF-binding heparan sulfate proteoglycan and the variant surface glycoprotein (a model GPI-anchored protein) from BM cultures. An involvement of proteases in this process is unlikely, because released proteoglycan contained the GPI anchor component, ethanol-amine, and protease inhibitors did not diminish the release. The mechanism of release is likely to involve a GPI-PLD and not a GPI-specific phospholipase C, because the release of variant surface glycoprotein did not reveal an epitope called the cross-reacting determinant that is exposed by phospholipase C-catalyzed GPI anchor cleavage. In addition, phosphatidic acid (which is specifically a product of GPI-PLD-catalyzed anchor cleavage) was generated during the spontaneous release of the GPI-anchored variant surface glycoprotein. We also detected GPI-PLD-specific enzyme activity and mRNA in BM cells. Therefore, we conclude that an endogenous GPI-PLD releases bFGF-heparan sulfate proteoglycan complexes from human BM cultures. This mechanism of GPI anchor cleavage could be relevant for mobilizing biologically active bFGF in BM. An endogenous GPI-PLD could also release other GPI-anchored proteins important for hematopoiesis and other physiologic processes.


Subject(s)
Bone Marrow/metabolism , Fibroblast Growth Factor 2/metabolism , Glycosylphosphatidylinositols/physiology , Heparitin Sulfate/metabolism , Phospholipase D/physiology , Proteoglycans/metabolism , Adult , Cells, Cultured , Endopeptidases/physiology , Glycosylphosphatidylinositols/genetics , Hematopoiesis , Heparan Sulfate Proteoglycans , Humans , Phospholipase D/genetics
12.
EMBO J ; 13(7): 1741-51, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-7512501

ABSTRACT

Although many glycosylphosphatidylinositol (GPI)-anchored proteins have been observed as soluble forms, the mechanisms by which they are released from the cell surface have not been demonstrated. We show here that a cell-associated GPI-specific phospholipase D (GPI-PLD) releases the GPI-anchored, complement regulatory protein decay-accelerating factor (DAF) from HeLa cells, as well as the basic fibroblast growth factor-binding heparan sulfate proteoglycan from bone marrow stromal cells. DAF found in the HeLa cell culture supernatants contained both [3H]ethanolamine and [3H]inositol, but not [3H]palmitic acid, whereas the soluble heparan sulfate proteoglycan present in bone marrow stromal cell culture supernatants contained [3H]ethanolamine. 125I-labeled GPI-DAF incorporated into the plasma membranes of these two cell types was released in a soluble form lacking the fatty acid GPI-anchor component. GPI-PLD activity was detected in lysates of both HeLa and bone marrow stromal cells. Treatment of HeLa cells with 1,10-phenanthroline, an inhibitor of GPI-PLD, reduced the release of [3H]ethanolamine-DAF by 70%. The hydrolysis of these GPI-anchored molecules is likely to be mediated by an endogenous GPI-PLD because [3H]ethanolamine DAF is constitutively released from HeLa cells maintained in serum-free medium. Furthermore, using PCR, a GPI-PLD mRNA has been identified in cDNA libraries prepared from both cell types. These studies are the first demonstration of the physiologically relevant release of GPI-anchored proteins from cells by a GPI-PLD.


Subject(s)
Antigens, CD/metabolism , Glycosylphosphatidylinositols/metabolism , Heparitin Sulfate/metabolism , Membrane Glycoproteins/metabolism , Phospholipase D/metabolism , Proteoglycans/metabolism , Bone Marrow/enzymology , Bone Marrow Cells , CD55 Antigens , DNA, Complementary/genetics , Ethanolamine , Ethanolamines/metabolism , Gene Library , HeLa Cells/enzymology , Heparan Sulfate Proteoglycans , Humans , Inositol/metabolism , Phenanthrolines/pharmacology , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , RNA, Messenger/genetics
13.
Blood ; 83(4): 907-10, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-7509211

ABSTRACT

Stem cell factor (SCF) and basic fibroblast growth factor (bFGF) are hematopoietic cytokines produced by bone marrow stromal cells. It is known that, although SCF and bFGF have limited clonogenic activity on their own, they can augment colony-stimulating factor (CSF)-mediated progenitor cell growth. Because these factors are both sequestered by stromal cells, we examined their interaction on progenitor cell growth in conjunction with granulocyte-macrophage-CSF (GM-CSF). In this study, we show that clonogenic growth derived from low-density bone marrow cells stimulated by GM-CSF is significantly augmented (P < .001) in the presence of maximal (100 ng/mL) concentrations of SCF in combination with 100 ng/mL of bFGF. When CD34+ cells are used, the synergistic effect of bFGF and SCF for GM-CSF-mediated progenitor cell growth is further increased, resulting in as much as a sevenfold increase in detectable colony-forming units granulocyte-macrophage (P < .001). These data suggest that the synergistic activity of bFGF and SCF is mediated directly on hematopoietic precursors. These observations suggest that bFGF and SCF, concentrated locally on stromal cell surfaces, might interact in concert with other hematopoietic cytokines to regulate stem cell proliferation and differentiation in hematopoietic niches in the bone marrow.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/cytology , Antigens, CD/analysis , Antigens, CD34 , Bone Marrow Cells , Cell Adhesion Molecules/pharmacology , Cell Division/drug effects , Drug Synergism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/pharmacology , Recombinant Proteins/pharmacology , Stem Cell Factor
14.
Blood ; 82(2): 430-5, 1993 Jul 15.
Article in English | MEDLINE | ID: mdl-8329701

ABSTRACT

Basic fibroblast growth factor (bFGF), a multifunctional growth factor produced by bone marrow stromal cells, is known to be a potent modulator of hematopoiesis. Because bFGF is present in both human megakaryocytes (MKs) and platelets, we have hypothesized that this growth factor might affect human megakaryocytopoiesis. To test this hypothesis, either low density bone marrow (BM) cells (LDBM), a human BM subpopulation (CD34+ DR+) enriched for the colony-forming unit megakaryocyte (CFU-MK) or a BM subpopulation (CD34+ DR-) enriched for the more primitive burst-forming unit megakaryocyte (BFU-MK) were assayed in the presence of this growth factor. The effect of bFGF on MK colony formation differed according to the cell population assayed. bFGF alone had on MK colony-stimulating activity (MK-CSA) when either CD34+ DR+ or CD34+ DR- BM cells were cloned, but exhibited MK-CSA equivalent to that of interleukin-3 (IL-3) when LDBM cells were used as the target cell population. The MK-CSA of bFGF was inhibited by the addition of neutralizing antisera to either IL-3 and/or granulocyte-macrophage colony-stimulating factor (GM-CSF) but not IL-6. The addition of excess amounts of either IL-3 or GM-CSF to cultures containing bFGF plus anti-IL-3 or anti-GM-CSF reversed the inhibition by the corresponding antisera. The addition of bFGF and IL-3 to assays containing CD34+ DR+ or CD34+ DR- cells increased the size of both CFU-MK- and BFU-MK-derived colonies, respectively, when compared with assays containing IL-3 alone. This increase in MK colony size mediated by bFGF was not affected by addition of either an anti-GM-CSF or anti-IL-6 neutralizing antisera. When LDBM cells were assayed, bFGF alone increased CFU-MK-derived colony size when compared with control values. However, this potentiation of MK colony size by bFGF could be reversed by the addition of either anti-IL-3 or anti-GM-CSF but not anti-IL-6 antisera. In addition, the effects of bFGF and IL-3 on the size of MK colonies cloned from LDBM were not additive. These results suggest that bFGF affects human megakaryocytopoiesis by directly promoting MK progenitor cell proliferation and stimulating BM accessory cells to release growth factor(s) with MK-CSA, such as IL-3 and GM-CSF. We conclude that bFGF, likely produced by cellular components of the BM microenvironment, plays an important role in the control of human megakaryocytopoiesis.


Subject(s)
Bone Marrow Cells , Fibroblast Growth Factor 2/pharmacology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Antibodies , Bone Marrow/drug effects , Cell Division , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/immunology , Interleukin-3/pharmacology , Interleukin-6/immunology , Megakaryocytes/drug effects , Platelet Count , Recombinant Proteins/pharmacology
15.
J Cell Physiol ; 155(3): 568-78, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7684043

ABSTRACT

Recently, a novel class of angiostatic steroids which block angiogenesis in several systems has been described. Since the elaboration of proteases is believed to be an important component of angiogenesis, we tested whether these steroids blocked the fibrinolytic response of endothelial cells to the angiogenic protein, basic fibroblast growth factor [bFGF]). Cultured bovine aortic endothelial (BAE) cells were incubated with bFGF and/or medroxyprogesterone acetate (MPA), an angiostatic steroid which has been shown to inhibit vascularization, collagenolysis, and tumor growth. When bFGF (3 ng/ml) was added to confluent monolayers of BAE cells, plasminogen activator (PA) activity in the medium was increased threefold. In contrast, MPA at 10(-6) M, 10(-7) M, 10(-8) M, and 10(-9) M decreased PA levels in the medium by 83%, 83%, 75%, and 39%, respectively. The stimulation of PA levels in BAE cells by bFGF (3 ng/ml) was abrogated by the presence of 10(-6) M MPA. This decrease in PA activity was found to be mediated by a significant increase in plasminogen activator inhibitor type-1 (PAI-1) production. MPA, therefore, negated one of the important enzymatic activities associated with the angiogenic process. In contrast to the decreased levels of secreted PA in cultures exposed simultaneously to MPA and bFGF, cell-associated PA levels remained high, consistent with earlier observations indicating that PAI-1 does not inhibit cell-associated PA. Thus, angiostatic steroids may exert their inhibitory effects on angiogenesis by increasing the synthesis of PAI-1. This, in turn, inhibits PA activity and, therefore, plasmin generation, which is essential for the invasive aspect of angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Pathologic , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activators/antagonists & inhibitors , Steroids/pharmacology , 17-alpha-Hydroxyprogesterone , Animals , Aorta , Cattle , Cells, Cultured , Dexamethasone/pharmacology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Heparin/pharmacology , Hydrocortisone/pharmacology , Hydroxyprogesterones/pharmacology , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/pharmacology , Plasminogen Activators/metabolism , Urokinase-Type Plasminogen Activator/metabolism
16.
Blood ; 81(3): 631-8, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-8427958

ABSTRACT

We have shown previously that basic fibroblast growth factor (bFGF) is a mitogen for human bone marrow (BM) stromal cells and that bFGF stimulates myelopoiesis in primary BM cultures. In this article, we demonstrate the presence of bFGF in two cell lineages in human BM and peripheral blood as well as the deposition of bFGF into the extracellular matrix of BM stromal cell cultures. In immunofluorescence experiments on BM and peripheral blood smears, megakaryocytes and platelets stained strongly for bFGF, whereas weaker staining was observed in immature and mature cells of the granulocyte series. The presence of bFGF in platelets was confirmed by enzyme-linked immunosorbent assay as well as by immunoprecipitation followed by immunoblotting. bFGF was synthesized by BM stromal cell cultures and was found either cell associated or localized in the nucleus and the nucleoli, and its location was dependent on the fixation procedure used. Addition of exogenous bFGF to stromal cells showed the presence of extracellular binding molecules for this cytokine. bFGF could be released from these sites by soluble heparin or phosphatidylinositol-specific phospholipase C. This study supports the role of bFGF as a stromal cell mitogen and stimulator of myelopoiesis. The data indicate that the stromal cells produce bFGF and that their extracellular matrix can serve as a reservoir for this growth factor. In addition, the results suggest a possible involvement of bFGF in platelet function as well as in megakaryocytopoiesis.


Subject(s)
Blood Platelets/physiology , Bone Marrow/physiology , Fibroblast Growth Factor 2/biosynthesis , Hematopoietic Stem Cells/physiology , Antibodies, Monoclonal , Bone Marrow Cells , Cells, Cultured , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/isolation & purification , Fluorescent Antibody Technique , Hematopoietic Stem Cells/cytology , Immunoblotting , In Vitro Techniques , Molecular Weight
17.
Blood ; 81(4): 909-15, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-8427999

ABSTRACT

We have previously shown that basic fibroblast growth factor (bFGF) is mitogenic for human bone marrow stromal cells and enhances myelopoiesis in human long-term bone marrow culture. In the present study, we examined the mechanism by which bFGF enhances granulopoiesis. We observed that bFGF significantly abrogated the inhibitory effect of transforming growth factor-beta 1 (TGF-beta 1) on granulocyte-macrophage colony-stimulating factor (GM-CSF)-supported progenitor cell growth (P = .009). The partial reversal of TGF-beta 1-mediated suppression was dependent on the dose of bFGF used. In addition, we noted that the inclusion of neutralizing antibody to TGF-beta 1 significantly augmented the clonogenic response to GM-CSF. We have also shown that 10 ng/mL or 100 ng/mL of bFGF resulted in a 30% to 100% increase in GM-CSF-mediated progenitor cell growth (P = .0001). These data suggest that bFGF may enhance myelopoiesis by modulating the inhibitory response to TGF-beta 1.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Transforming Growth Factor beta/pharmacology , Antibodies , Cell Division , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunization, Passive , Transforming Growth Factor beta/immunology
18.
Int J Cancer ; 51(5): 788-91, 1992 Jul 09.
Article in English | MEDLINE | ID: mdl-1612785

ABSTRACT

Two human melanoma cell lines, UCT-Mel 2 and UCT-Mel 3, were invariably tumorigenic in nude mice when inoculated s.c. in doses of 10(6) cells or higher; 10(5) cells or less did not give rise to tumours. In this report we show that otherwise sub-tumorigenic inocula developed into vigorously growing tumour xenografts when co-inoculated with normal fibroblasts. Fibroblasts derived from adult, neonatal or embryonic tissues all functioned as complementing cells, as did cells of human or murine origin. There was, however, a requirement for complementing cell viability, since ethanol-killed fibroblasts were inefficacious. The fibroblast effect was dose-dependent and was not observed if injections of fibroblasts and melanoma cells were separated anatomically or temporally. We have shown, by titrating admixtures of melanoma cells and fibroblasts, that fibroblasts are, in quantitative terms, more efficacious than melanoma cells as complementing cells. The system we describe provides a useful model for the study of stromal-cell regulation of tumour growth.


Subject(s)
Fibroblasts/physiology , Melanoma, Experimental/pathology , Animals , Cell Count , Culture Media , Ethanol/pharmacology , Fibroblasts/drug effects , Fibroblasts/transplantation , Humans , Kinetics , Mice , Mice, Nude , Neoplasm Transplantation , Tumor Cells, Cultured
19.
Br J Cancer ; 65(4): 487-90, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1562456

ABSTRACT

When human melanoma cells are injected into nude mice they usually give rise to tumours that grow progressively and do not elicit a prominent host response. We have recently developed a melanoma cell line, UCT-Mel 7, that did not show these characteristics. In the first place UCT-Mel 7 showed a consistently unusual, phasic growth pattern. After a short initial period of limited growth (phase 1), the tumour ceased growing and remained static for 2-3 months (phase 2). The tumour then regressed (phase 3) to enter a second period of quiescence (phase 4) which was eventually broken by the emergence of a rapidly growing lethal tumour (phase 5). Of particular interest was the fact that the rate at which the tumours grew correlated closely with their collagen content. During the prolonged, phase 2 plateau, the tumours were intensely desmoplastic; rapidly growing phase 5 tumours, that had escaped from dormancy, contained very little collagen and virtually no reticulin. This cell line helps to fill an important need for an experimental system for the study of desmoplasia, dormancy and progression.


Subject(s)
Melanoma/pathology , Animals , Collagen/metabolism , Histocytochemistry , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Reticulin/metabolism , Tumor Cells, Cultured
20.
Blood ; 79(5): 1178-84, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1536945

ABSTRACT

Plasminogen activators (PAs) and/or plasmin may be involved in hematopoietic regulation. These enzymes release biologically relevant cytokines such as basic fibroblast growth factor (bFGF) from matrix and cell surfaces. In addition, transforming growth factor beta (TGF beta) and interleukin-1 beta (IL-1 beta) are converted from inactive to active forms by plasmin. Therefore, we studied the regulation of PAs and their specific inhibitors, PA inhibitor 1 (PAI-1) and PA inhibitor 2 (PAI-2), in human bone marrow stromal fibroblasts by IL-1 beta, bFGF, and TGF beta. All three cytokines stimulated PA secretion. IL-1 beta at 10(4) U/mL increased urokinase (u-PA) levels approximately 10-fold, bFGF at 0.2 ng/mL also increased production 10-fold, but increased predominantly tissue PA (t-PA) expression. TGF beta at 0.2 ng/mL increased u-PA production up to 300-fold. PAI-1 and PAI-2 are also regulated by these cytokines. IL-1 beta decreased PAI-1 levels by 50% and stimulated PAI-2 levels sixfold. bFGF had minimal effects on PAI-1 and TGF beta increased PAI-1 levels twofold. Neither of these agents had an effect on PAI-2 levels. Thus, three cytokines relevant to bone marrow physiology regulate PA and inhibitor production by human bone marrow stromal fibroblasts. In this manner PA and plasmin generation in specific microenvironments in the bone marrow may be one of the factors orchestrating the complex series of events, which results in an efficient exquisitely regulated hematopoietic process.


Subject(s)
Bone Marrow/enzymology , Fibroblast Growth Factor 2/pharmacology , Interleukin-1/pharmacology , Plasminogen Activators/metabolism , Plasminogen Inactivators/metabolism , Transforming Growth Factor beta/pharmacology , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Fibroblasts/enzymology , Humans , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL